Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Bottlenecks in HIV-1 transmission: insights from the study of founder viruses

Key Points

  • Approximately 80% of HIV-1 heterosexual transmission events are established from a single transmitted/founder (T/F) virus.

  • During transmission, multiple bottlenecks reduce the viral population from the large, genetically variable population in the blood of a chronically infected donor to a single T/F virus in the newly infected recipient.

  • Recent evidence suggests that transmission of a single T/F virus results from both stochastic bottlenecks that restrict transmission of all viruses (for example, nonspecific barrier functions) and selective bottlenecks that favour transmission of viruses with specific phenotypes.

  • Understanding the bottlenecks that restrict transmission may inform targets for prevention strategies. For example, antiretrovirals can be used to suppress the viral population in infected individuals and prevent them from sexually transmitting HIV-1 (that is, 'treatment as prevention').

Abstract

HIV-1 infection typically results from the transmission of a single viral variant, the transmitted/founder (T/F) virus. Studies of these HIV-1 variants provide critical information about the transmission bottlenecks and the selective pressures acting on the virus in the transmission fluid and in the recipient tissues. These studies reveal that T/F virus phenotypes are shaped by stochastic and selective forces that restrict transmission and may be targets for prevention strategies. In this Review, we highlight how studies of T/F viruses contribute to a better understanding of the biology of HIV-1 transmission and discuss how these findings affect HIV-1 prevention strategies.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The transmitted/founder virus is shaped by multiple genetic bottlenecks.
Figure 2: Compartmentalization of genital tract-specific viral lineages.
Figure 3: Selection of transmitted/founder virus phenotypes.
Figure 4: Characteristics of transmitted/founder viruses.

Similar content being viewed by others

References

  1. Derdeyn, C. A. et al. Envelope-constrained neutralization-sensitive HIV-1 after heterosexual transmission. Science 303, 2019–2022 (2004).

    CAS  PubMed  Google Scholar 

  2. Abrahams, M. R. et al. Quantitating the multiplicity of infection with human immunodeficiency virus type 1 subtype C reveals a non-poisson distribution of transmitted variants. J. Virol. 83, 3556–3567 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Keele, B. F. et al. Identification and characterization of transmitted and early founder virus envelopes in primary HIV-1 infection. Proc. Natl Acad. Sci. USA 105, 7552–7557 (2008). One of the first studies using single genome amplification to amplify env genes from blood samples collected during acute infection and infer the T/F sequence and T/F virus phenotypes. It found that 76% of sexual transmission events were established from a single T/F virus and that 98% were CCR5 tropic.

    CAS  PubMed  Google Scholar 

  4. Haaland, R. E. et al. Inflammatory genital infections mitigate a severe genetic bottleneck in heterosexual transmission of subtype A and C HIV-1. PLoS Pathog. 5, e1000274 (2009).

    PubMed  PubMed Central  Google Scholar 

  5. Fieni, F. et al. Viral RNA levels and env variants in semen and tissues of mature male rhesus macaques infected with SIV by penile inoculation. PLoS ONE 8, e76367 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Chaillon, A. et al. HIV migration between blood and cerebrospinal fluid or semen over time. J. Infect. Dis. 209, 1642–1652 (2014).

    PubMed  Google Scholar 

  7. Anderson, J. A. et al. HIV-1 populations in semen arise through multiple mechanisms. PLoS Pathog. 6, e1001053 (2010). This study illustrates that diverse, compartmentalized viral lineages can be detected in some semen samples.

    PubMed  PubMed Central  Google Scholar 

  8. Boeras, D. I. et al. Role of donor genital tract HIV-1 diversity in the transmission bottleneck. Proc. Natl Acad. Sci. USA 108, E1156–E1163 (2011). A study of transmission pairs suggesting that the transmission bottleneck is not just stochastic and may select for rare variants in the donor GT.

    CAS  PubMed  Google Scholar 

  9. Byrn, R. A. & Kiessling, A. A. Analysis of human immunodeficiency virus in semen: indications of a genetically distinct virus reservoir. J. Reprod. Immunol. 41, 161–176 (1998).

    CAS  PubMed  Google Scholar 

  10. Delwart, E. L. et al. Human immunodeficiency virus type 1 populations in blood and semen. J. Virol. 72, 617–623 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Diem, K. et al. Male genital tract compartmentalization of human immunodeficiency virus type 1 (HIV). AIDS Res. Hum. Retroviruses 24, 561–571 (2008).

    CAS  PubMed  Google Scholar 

  12. Gupta, P. et al. Human immunodeficiency virus type 1 shedding pattern in semen correlates with the compartmentalization of viral quasi species between blood and semen. J. Infect. Dis. 182, 79–87 (2000).

    CAS  PubMed  Google Scholar 

  13. Kiessling, A. A. et al. Human immunodeficiency virus in semen arises from a genetically distinct virus reservoir. AIDS Res. Hum. Retroviruses 14, S33–S41 (1998).

    PubMed  Google Scholar 

  14. Overbaugh, J., Anderson, R. J., Ndinya-Achola, J. O. & Kreiss, J. K. Distinct but related human immunodeficiency virus type 1 variant populations in genital secretions and blood. AIDS Res. Hum. Retroviruses 12, 107–115 (1996).

    CAS  PubMed  Google Scholar 

  15. Zhu, T. F. et al. Genetic characterization of human immunodeficiency virus type 1 in blood and genital secretions: evidence for viral compartmentalization and selection during sexual transmission. J. Virol. 70, 3098–3107 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Bull, M. et al. Compartmentalization of HIV-1 within the female genital tract is due to monotypic and low-diversity variants not distinct viral populations. PLoS ONE 4, e7122 (2009).

    PubMed  PubMed Central  Google Scholar 

  17. Poss, M. et al. Evolution of envelope sequences from the genital tract and peripheral blood of women infected with clade A human immunodeficiency virus type 1. J. Virol. 72, 8240–8251 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Bull, M. E. et al. Human immunodeficiency viruses appear compartmentalized to the female genital tract in cross-sectional analyses but genital lineages do not persist over time. J. Infect. Dis. 207, 1206–1215 (2013).

    PubMed  PubMed Central  Google Scholar 

  19. Bull, M. E. et al. Monotypic human immunodeficiency virus type 1 genotypes across the uterine cervix and in blood suggest proliferation of cells with provirus. J. Virol. 83, 6020–6028 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Roberts, L. et al. Genital tract inflammation during early HIV-1 infection predicts higher plasma viral load set point in women. J. Infect. Dis. 205, 194–203 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Butler, D. M. et al. The origins of sexually transmitted HIV among men who have sex with men. Sci. Transl Med. 2, 18re1 (2010).

    PubMed  PubMed Central  Google Scholar 

  22. Heath, L., Frenkel, L. M., Foley, B. T. & Mullins, J. I. Comment on “The origins of sexually transmitted HIV among men who have sex with men”. Sci. Transl Med. 2, 50le1 (2010).

    PubMed  PubMed Central  Google Scholar 

  23. Gianella, S. et al. Sexual transmission of predicted CXCR4-tropic HIV-1 likely originating from the source partner's seminal cells. Virology 434, 2–4 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Salle, B. et al. Infection of macaques after vaginal exposure to cell-associated simian immunodeficiency virus. J. Infect. Dis. 202, 337–344 (2010).

    PubMed  Google Scholar 

  25. Miller, C. J. et al. Genital mucosal transmission of simian immunodeficiency virus: animal model for heterosexual transmission of human immunodeficiency virus. J. Virol. 63, 4277–4284 (1989).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Gnanakaran, S. et al. Recurrent signature patterns in HIV-1 B clade envelope glycoproteins associated with either early or chronic infections. PLoS Pathog. 7, e1002209 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Chohan, B. et al. Selection for human immunodeficiency virus type 1 envelope glycosylation variants with shorter V1-V2 loop sequences occurs during transmission of certain genetic subtypes and may impact viral RNA levels. J. Virol. 79, 6528–6531 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Sagar, M. et al. Selection of HIV variants with signature genotypic characteristics during heterosexual transmission. J. Infect. Dis. 199, 580–589 (2009).

    PubMed  PubMed Central  Google Scholar 

  29. Ping, L. H. et al. Comparison of viral Env proteins from acute and chronic infections with subtype C human immunodeficiency virus type 1 identifies differences in glycosylation and CCR5 utilization and suggests a new strategy for immunogen design. J. Virol. 87, 7218–7233 (2013). This study of a large number of heterosexually transmitted T/F env genes found that they are exclusively T cell-tropic. This study also observed that, relative to their controls, T/F viruses isolated from males have fewer glycosylation sites than T/F viruses isolated from females. This suggests that female-to-male transmission either selects viruses with fewer glycosylation sites or selects viruses lacking specific glycosylation sites.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Frost, S. D. et al. Characterization of human immunodeficiency virus type 1 (HIV-1) envelope variation and neutralizing antibody responses during transmission of HIV-1 subtype B. J. Virol. 79, 6523–6527 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Liu, Y. et al. Env length and N-linked glycosylation following transmission of human immunodeficiency virus Type 1 subtype B viruses. Virology 374, 229–233 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Curlin, M. E. et al. HIV-1 envelope subregion length variation during disease progression. PLoS Pathog. 6, e1001228 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Sagar, M., Wu, X., Lee, S. & Overbaugh, J. Human immunodeficiency virus type 1 V1-V2 envelope loop sequences expand and add glycosylation sites over the course of infection, and these modifications affect antibody neutralization sensitivity. J. Virol. 80, 9586–9598 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Russell, E. S. et al. The genetic bottleneck in vertical transmission of subtype C HIV-1 is not driven by selection of especially neutralization-resistant virus from the maternal viral population. J. Virol. 85, 8253–8262 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Wei, X. et al. Antibody neutralization and escape by HIV-1. Nature 422, 307–312 (2003).

    CAS  PubMed  Google Scholar 

  36. West, A. P. et al. Structural insights on the role of antibodies in HIV-1 vaccine and therapy. Cell 156, 633–648 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Wilen, C. B. et al. Phenotypic and immunologic comparison of clade B transmitted/founder and chronic HIV-1 envelope glycoproteins. J. Virol. 85, 8514–8527 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Lavine, C. L. et al. High-mannose glycan-dependent epitopes are frequently targeted in broad neutralizing antibody responses during human immunodeficiency virus type 1 infection. J. Virol. 86, 2153–2164 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Balzarini, J. Targeting the glycans of glycoproteins: a novel paradigm for antiviral therapy. Nat. Rev. Microbiol. 5, 583–597 (2007).

    CAS  PubMed  Google Scholar 

  40. Ballegaard, V., Haugaard, A. K., Garred, P., Nielsen, S. D. & Munthe-Fog, L. The lectin pathway of complement: advantage or disadvantage in HIV pathogenesis? Clin. Immunol. 154, 13–25 (2014).

    CAS  PubMed  Google Scholar 

  41. Adamek, M. et al. Characterization of mannose-binding lectin (MBL) variants by allele-specific sequencing of MBL2 and determination of serum MBL protein levels. Tissue Antigens 82, 410–415 (2013).

    CAS  PubMed  Google Scholar 

  42. Wing, J. B. et al. Mannose-binding lectin is present in human semen and modulates cellular adhesion of Neisseria gonorrhoeae in vitro. Clin. Exp. Immunol. 157, 408–414 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Munch, J. et al. Semen-derived amyloid fibrils drastically enhance HIV infection. Cell 131, 1059–1071 (2007).

    PubMed  Google Scholar 

  44. Kim, K. A. et al. Semen-mediated enhancement of HIV infection is donor-dependent and correlates with the levels of SEVI. Retrovirology 7, 1–12 (2010).

    CAS  Google Scholar 

  45. Roan, N. R. et al. Peptides released by physiological cleavage of semen coagulum proteins form amyloids that enhance HIV infection. Cell Host Microbe 10, 541–550 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Gulick, R. M. et al. Maraviroc for previously treated patients with R5 HIV-1 infection. New Engl. J. Med. 359, 1429–1441 (2008).

    CAS  PubMed  Google Scholar 

  47. Zirafi, O. et al. Semen-mediated enhancement of HIV infection markedly impairs the antiviral efficacy of microbicides. AIDS Res. Hum. Retroviruses 30, A183–A183 (2014).

    Google Scholar 

  48. Munch, J. et al. Effect of semen and seminal amyloid on vaginal transmission of simian immunodeficiency virus. Retrovirology 10, 1–9 (2013).

    Google Scholar 

  49. Cohen, M. S. et al. Prevention of HIV-1 infection with early antiretroviral therapy. New Engl. J. Med. 365, 493–505 (2011). This study showed that early antiretroviral therapy reduced transmission rates by 96%.

    CAS  PubMed  Google Scholar 

  50. Quinn, T. C. et al. Viral load and heterosexual transmission of human immunodeficiency virus type 1. New Engl. J. Med. 342, 921–929 (2000).

    CAS  PubMed  Google Scholar 

  51. Baeten, J. M. et al. Genital HIV-1 RNA predicts risk of heterosexual HIV-1 transmission. Sci. Transl Med. 3, 77ra29 (2011).

    PubMed  PubMed Central  Google Scholar 

  52. Butler, D. A. et al. Herpes simplex virus 2 serostatus and viral loads of HIV-1 in blood and semen as risk factors for HIV transmission among men who have sex with men. AIDS 22, 1667–1671 (2008).

    PubMed  PubMed Central  Google Scholar 

  53. Cohen, M. S. Sexually transmitted diseases enhance HIV transmission: no longer a hypothesis. Lancet 351, 5–7 (1998).

    PubMed  Google Scholar 

  54. Moss, W. J. et al. Suppression of human immunodeficiency virus replication during acute measles. J. Infect. Dis. 185, 1035–1042 (2002).

    PubMed  Google Scholar 

  55. Watt, G., Kantipong, P. & Jongsakul, K. Decrease in human immunodeficiency virus type 1 load during acute dengue fever. Clin. Infect. Dis. 36, 1067–1069 (2003).

    PubMed  Google Scholar 

  56. Anzala, A. O. et al. Acute sexually transmitted infections increase human immunodeficiency virus type 1 plasma viremia, increase plasma type 2 cytokines, and decrease CD4 cell counts. J. Infect. Dis. 182, 459–466 (2000).

    CAS  PubMed  Google Scholar 

  57. Cohen, M. S. et al. Reduction of concentration of HIV-1 in semen after treatment of urethritis: implications for prevention of sexual transmission of HIV-1. Lancet 349, 1868–1873 (1997).

    CAS  PubMed  Google Scholar 

  58. Malott, R. J. et al. Neisseria gonorrhoeae-derived heptose elicits an innate immune response and drives HIV-1 expression. Proc. Natl Acad. Sci. USA 110, 10234–10239 (2013).

    CAS  PubMed  Google Scholar 

  59. Nagot, N. et al. Reduction of HIV-1 RNA levels with therapy to suppress herpes simplex virus. New Engl. J. Med. 356, 790–799 (2007).

    CAS  PubMed  Google Scholar 

  60. Nixon, B. et al. Genital herpes simplex virus type 2 infection in humanized HIV-transgenic mice triggers HIV shedding and is associated with greater neurological disease. J. Infect. Dis. 209, 510–522 (2014).

    CAS  PubMed  Google Scholar 

  61. Wilen, C. B., Tilton, J. C. & Doms, R. W. in HIV: From Biology to Prevention and Treatment (eds Bushman, F. D., Nabel, G. J. & Swanstrom, R.) (Cold Spring Harbor Laboratory, 2011).

    Google Scholar 

  62. Berger, E. A., Murphy, P. M. & Farber, J. M. Chemokine receptors as HIV-1 coreceptors: roles in viral entry, tropism, and disease. Annu. Rev. Immunol. 17, 657–700 (1999).

    CAS  PubMed  Google Scholar 

  63. Johnston, S. H. et al. A quantitative affinity-profiling system that reveals distinct CD4/CCR5 usage patterns among human immunodeficiency virus type 1 and simian immunodeficiency virus strains. J. Virol. 83, 11016–11026 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Joseph, S. B. et al. Quantification of entry phenotypes of macrophage-tropic HIV-1 across a wide range of CD4 densities. J. Virol. 88, 1858–1869 (2014).

    PubMed  PubMed Central  Google Scholar 

  65. Parrish, N. F. et al. Transmitted/founder and chronic subtype C HIV-1 use CD4 and CCR5 receptors with equal efficiency and are not inhibited by blocking the integrin α4β7 . PLoS Pathog. 8, e1002686 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Isaacman-Beck, J. et al. Heterosexual transmission of human immunodeficiency virus type 1 subtype C: macrophage tropism, alternative coreceptor use, and the molecular anatomy of CCR5 utilization. J. Virol. 83, 8208–8220 (2009). The first study illustrating that T/F viruses are not macrophage-tropic and that transmission does not select for enhanced macrophage-tropism.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Li, H. et al. High multiplicity infection by HIV-1 in men who have sex with men. PLoS Pathog. 6, e1000890 (2010).

    PubMed  PubMed Central  Google Scholar 

  68. Ochsenbauer, C. et al. Generation of transmitted/founder HIV-1 infectious molecular clones and characterization of their replication capacity in CD4 T lymphocytes and monocyte-derived macrophages. J. Virol. 86, 2715–2728 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  69. Salazar-Gonzalez, J. F. et al. Genetic identity, biological phenotype, and evolutionary pathways of transmitted/founder viruses in acute and early HIV-1 infection. J. Exp. Med. 206, 1273–1289 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Parker, Z. F. et al. Transmitted/founder and chronic HIV-1 envelope proteins are distinguished by differential utilization of CCR5. J. Virol. 87, 2401–2411 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Parrish, N. F. et al. Phenotypic properties of transmitted founder HIV-1. Proc. Natl Acad. Sci. USA 110, 6626–6633 (2013). A Thorough analysis of T/F virus phenotypes, which observed that subtype B T/F viruses are more resistant to IFNα than controls.

    CAS  PubMed  Google Scholar 

  72. Asmal, M. et al. A signature in HIV-1 envelope leader peptide associated with transition from acute to chronic infection impacts envelope processing and infectivity. PLoS ONE 6, e23673 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Banchereau, J. & Steinman, R. M. Dendritic cells and the control of immunity. Nature 392, 245–252 (1998).

    CAS  PubMed  Google Scholar 

  74. Shen, R. et al. Vaginal myeloid dendritic cells transmit founder HIV-1. J. Virol. 88, 7683–7688 (2014).

    PubMed  PubMed Central  Google Scholar 

  75. Cameron, P. U. et al. Dendritic cells exposed to human immunodeficiency virus type-1 transmit a vigorous cytopathic infection to CD4+ T cells. Science 257, 383–386 (1992).

    CAS  PubMed  Google Scholar 

  76. Pope, M. et al. Conjugates of dendritic cells and memory T lymphocytes from skin facilitate productive infection with HIV-1. Cell 78, 389–398 (1994).

    CAS  PubMed  Google Scholar 

  77. Kawamura, T. et al. Candidate microbicides block HIV-1 infection of human immature Langerhans cells within epithelial tissue explants. J. Exp. Med. 192, 1491–1500 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Kawamura, T. et al. R5 HIV productively infects Langerhans cells, and infection levels are regulated by compound CCR5 polymorphisms. Proc. Natl Acad. Sci. USA 100, 8401–8406 (2003).

    CAS  PubMed  Google Scholar 

  79. Tenner-Racz, K. et al. The unenlarged lymph nodes of HIV-1-infected, asymptomatic patients with high CD4 T cell counts are sites for virus replication and CD4 T cell proliferation, the impact of highly active antiretroviral therapy. J. Exp. Med. 187, 949–959 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  80. van der Ende, M. E. et al. CD4 T cells remain the major source of HIV-1 during end stage disease. AIDS 13, 1015–1019 (1999).

    CAS  PubMed  Google Scholar 

  81. Piguet, V. & Steinman, R. M. The interaction of HIV with dendritic cells: outcomes and pathways. Trends Immunol. 28, 503–510 (2007).

    CAS  PubMed  Google Scholar 

  82. Zhang, Z. Q. et al. Sexual transmission and propagation of SIV and HIV in resting and activated CD4+ T cells. Science 286, 1353–1357 (1999).

    CAS  PubMed  Google Scholar 

  83. Zhang, Z. Q. et al. Roles of substrate availability and infection of resting and activated CD4+ T cells in transmission and acute simian immunodeficiency virus infection. Proc. Natl Acad. Sci. USA 101, 5640–5645 (2004).

    CAS  PubMed  Google Scholar 

  84. Li, Q. et al. Glycerol monolaurate prevents mucosal SIV transmission. Nature 458, 1034–1038 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Atashili, J., Poole, C., Ndumbe, P. M., Adimora, A. A. & Smith, J. S. Bacterial vaginosis and HIV acquisition: a meta-analysis of published studies. AIDS 22, 1493–1501 (2008).

    PubMed  PubMed Central  Google Scholar 

  86. Freeman, E. E. et al. Herpes simplex virus 2 infection increases HIV acquisition in men and women: systematic review and meta-analysis of longitudinal studies. AIDS 20, 73–83 (2006).

    PubMed  Google Scholar 

  87. Carlson, J. M. et al. HIV transmission. Selection bias at the heterosexual HIV-1 transmission bottleneck. Science 345, 1254031 (2014). Shows that the severe genetic bottleneck at the time of transmission selects for high-fitness viruses.

    PubMed  PubMed Central  Google Scholar 

  88. Boutwell, C. L. et al. Frequent and variable cytotoxic-T-lymphocyte escape-associated fitness costs in the human immunodeficiency virus type 1 subtype B Gag proteins. J. Virol. 87, 3952–3965 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Martinez-Picado, J. et al. Fitness cost of escape mutations in p24 Gag in association with control of human immunodeficiency virus type 1. J. Virol. 80, 3617–3623 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  90. Schneidewind, A. et al. Escape from the dominant HLA-B27-restricted cytotoxic T-lymphocyte response in Gag is associated with a dramatic reduction in human immunodeficiency virus type 1 replication. J. Virol. 81, 12382–12393 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Allen, T. M. et al. Selection, transmission, and reversion of an antigen-processing cytotoxic T-lymphocyte escape mutation in human immunodeficiency virus type 1 infection. J. Virol. 78, 7069–7078 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Leslie, A. J. et al. HIV evolution: CTL escape mutation and reversion after transmission. Nat. Med. 10, 282–289 (2004).

    CAS  PubMed  Google Scholar 

  93. Li, B. et al. Rapid reversion of sequence polymorphisms dominates early human immunodeficiency virus type 1 evolution. J. Virol. 81, 193–201 (2007).

    CAS  PubMed  Google Scholar 

  94. Fischer, W. et al. Transmission of single HIV-1 genomes and dynamics of early immune escape revealed by ultra-deep sequencing. PLoS ONE 5, e12303 (2010).

    PubMed  PubMed Central  Google Scholar 

  95. Henn, M. R. et al. Whole genome deep sequencing of HIV-1 reveals the impact of early minor variants upon immune recognition during acute infection. PLoS Pathog. 8, e1002529 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Kijak, G. H. et al. Cryptic multiple HIV-1 infection revealed by early, frequent, and deep sampling during acute infection. AIDS Res. Hum. Retroviruses 30, A58 (2014).

    Google Scholar 

  97. Fenton-May, A. E. et al. Relative resistance of HIV-1 founder viruses to control by interferon-α. Retrovirology 10, 146 (2013).

    PubMed  PubMed Central  Google Scholar 

  98. Abel, K., Rocke, D. M., Chohan, B., Fritts, L. & Miller, C. J. Temporal and anatomic relationship between virus replication and cytokine gene expression after vaginal simian immunodeficiency virus infection. J. Virol. 79, 12164–12172 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Sandler, N. G. et al. Type I interferon responses in rhesus macaques prevent SIV infection and slow disease progression. Nature 511, 601–605 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  100. Nawaz, F. et al. The genotype of early-transmitting HIV gp120s promotes α4β7-reactivity, revealing α4β7+/CD4+ T cells as key targets in mucosal transmission. PLoS Pathog. 7, e1001301 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Wagner, N. et al. Critical role for β7 integrins in formation of the gut-associated lymphoid tissue. Nature 382, 366–370 (1996).

    CAS  PubMed  Google Scholar 

  102. Perez, L. G., Chen, H., Liao, H. X. & Montefiori, D. C. Envelope glycoprotein binding to the α4β7 integrin complex is not a general property of most HIV-1 variants. J. Virol. 88, 10767–10777 (2014).

    PubMed  PubMed Central  Google Scholar 

  103. Richardson, S. I. et al. The sequence of the α4β7-binding motif on gp120 of transmitted/founder viruses contributes to the dependence on the integrin for HIV infection. AIDS Res. Hum. Retroviruses 30, A56 (2014).

    Google Scholar 

  104. Byrareddy, S. N. et al. Targeting αβ integrin reduces mucosal transmission of simian immunodeficiency virus and protects gut-associated lymphoid tissue from infection. Nat. Med. 20, 1397–1400 (2014). This study illustrates that an antibody to α4β7 integrin can protect macaques from SIV acquisition.

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Schnell, G., Price, R. W., Swanstrom, R. & Spudich, S. Compartmentalization and clonal amplification of HIV-1 variants in the cerebrospinal fluid during primary infection. J. Virol. 84, 2395–2407 (2010).

    CAS  PubMed  Google Scholar 

  106. Sturdevant, C. B. et al. Central nervous system compartmentalization of HIV-1 subtype C variants early and late in infection in young children. PLoS Pathog. 8, e1003094 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  107. Del Prete, G. Q. et al. Molecularly tagged simian immunodeficiency virus SIVmac239 synthetic swarm for tracking independent infection events. J. Virol. 88, 8077–8090 (2014).

    PubMed  PubMed Central  Google Scholar 

  108. Coates, T. J., Richter, L. & Caceres, C. Behavioural strategies to reduce HIV transmission: how to make them work better. Lancet 372, 669–684 (2008).

    PubMed  PubMed Central  Google Scholar 

  109. Wetmore, C. M., Manhart, L. E. & Wasserheit, J. N. Randomized controlled trials of interventions to prevent sexually transmitted infections: learning from the past to plan for the future. Epidemiol. Rev. 32, 121–136 (2010).

    PubMed  PubMed Central  Google Scholar 

  110. Gray, R. H. et al. Male circumcision for HIV prevention in men in Rakai, Uganda: a randomised trial. Lancet 369, 657–666 (2007). These results of a randomized clinical trial show that circumcision significantly reduces the rate of HIV acquisition.

    PubMed  Google Scholar 

  111. Muessig, K. E. & Cohen, M. S. Advances in HIV prevention for serodiscordant couples. Curr. HIV/AIDS Rep. 11, 434–446 (2014).

    PubMed  PubMed Central  Google Scholar 

  112. Rodger, A. et al. HIV transmission risk through condomless sex if the HIV+ partner is on suppressive ART: PARTNER study. Top. Antivir. Med. 22, 24–25 (2014).

    Google Scholar 

  113. Smedley, J. et al. Tracking the luminal exposure and lymphatic drainage pathways of intravaginal and intrarectal inocula used in nonhuman primate models of HIV transmission. PLoS ONE 9, e92830 (2014).

    PubMed  PubMed Central  Google Scholar 

  114. Stieh, D. J. et al. Vaginal challenge with an SIV-based dual reporter system reveals that infection can occur throughout the upper and lower female reproductive tract. PLoS Pathog. 10, e1004440 (2014). This study demonstrates that cells throughout the reproductive tract of female macaques are susceptible to SIV.

    PubMed  PubMed Central  Google Scholar 

  115. Haynes, B. F. et al. Immune-correlates analysis of an HIV-1 vaccine efficacy trial. New Engl. J. Med. 366, 1275–1286 (2012).

    CAS  PubMed  Google Scholar 

  116. Rolland, M. et al. Increased HIV-1 vaccine efficacy against viruses with genetic signatures in Env V2. Nature 490, 417–420 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Rolland, M. et al. Genetic impact of vaccination on breakthrough HIV-1 sequences from the STEP trial. Nat. Med. 17, 366–371 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Roederer, M. et al. Immunological and virological mechanisms of vaccine-mediated protection against SIV and HIV. Nature 505, 502–508 (2014).

    CAS  PubMed  Google Scholar 

  119. Dolgin, E. Long-acting HIV drugs advanced to overcome adherence challenge. Nat. Med. 20, 323–324 (2014).

    CAS  PubMed  Google Scholar 

  120. Koharudin, L. M. I. & Gronenborn, A. M. Antiviral lectins as potential HIV microbicides. Curr. Opin. Virol. 7, 95–100 (2014).

    CAS  PubMed  Google Scholar 

  121. Ribeiro, R. M. et al. Estimation of the initial viral growth rate and basic reproductive number during acute HIV-1 infection. J. Virol. 84, 6096–6102 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  122. Mellors, J. W. et al. Prognosis in HIV-1 infection predicted by the quantity of virus in plasma. Science 272, 1167–1170 (1996).

    CAS  PubMed  Google Scholar 

  123. Fraser, C., Hollingsworth, T. D., Chapman, R., de Wolf, F. & Hanage, W. P. Variation in HIV-1 set-point viral load: epidemiological analysis and an evolutionary hypothesis. Proc. Natl Acad. Sci. USA 104, 17441–17446 (2007).

    CAS  PubMed  Google Scholar 

  124. Fraser, C. et al. Virulence and pathogenesis of HIV-1 infection: an evolutionary perspective. Science 343, 1243727 (2014).

    PubMed  PubMed Central  Google Scholar 

  125. Hodcroft, E. et al. The contribution of viral genotype to plasma viral set-point in HIV infection. PLoS Pathog. 10, e1004112 (2014).

    PubMed  PubMed Central  Google Scholar 

  126. Tang, J. M. et al. HLA allele sharing and HIV type 1 viremia in seroconverting Zambians with known transmitting partners. AIDS Res. Hum. Retroviruses 20, 19–25 (2004).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was funded by awards from the US National Institutes of Health (U01 AI067854 (CHAVI)), and R37 AI44667 to R.S. and R37 DK049381 to M.S.C.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Sarah B. Joseph.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

PowerPoint slides

Glossary

Retrovirus

A member of the family Retroviridae. Retroviruses use reverse transcriptase to convert their single-stranded RNA genome into double-stranded DNA, which is subsequently integrated into the host genome. The integrated proviral genome may be transcribed, translated and assembled into new virions.

Lentivirus

Genus of the family Retroviridae that includes HIV and simian immunodeficiency virus (SIV).

Transmitted/founder virus

(T/F virus). The virus that forms a systemic infection after being transferred from an infected individual to an uninfected individual. HIV-1 infections are typically established from a single T/F virus.

env

Gene encoding the HIV-1 Env glycoprotein. This protein is cleaved into two subunits (gp120 and gp41), which are non-covalently bound as heterodimers and organized as trimers on the virion surface. Env glycoproteins facilitate virus binding to and fusion with host cells.

Compartmentalized lineages

Virus variants found outside the blood that are genetically distinct from variants in the blood. Compartmentalized lineages evolve after many generations of independent replication in a tissue or compartment.

Transmission pairs

HIV-1 donors and the recipient that each infected. Studies of transmission pairs are useful for examining many aspects of transmission biology, including factors that influence the probability of transmission and whether transmission bottlenecks select for specific viral phenotypes.

Neutralizing antibodies

Antibodies that inactivate infectious agents after recognizing antigens on their surface.

Lectins

Carbohydrate-binding proteins capable of neutralizing viruses by binding glycans on the viral surface.

Glycosylation

The host process of attaching glycans to proteins. Glycosylation of HIV-1 Env has the effect of shielding epitopes on Env from antibody binding.

HIV subtypes

Group M HIV-1 contains multiple lineages, termed subtypes.

Autologous donor antibodies

Antibodies that are produced by the same individual who produced the viruses that they are neutralizing. May inhibit transmission by neutralizing viruses in the transmission fluid.

Heterologous antibodies

Antibodies that are produced by an individual different from the one who produced the viruses that they are neutralizing.

Pseudotyped viruses

Viruses produced in vitro using a method that allows the researcher to control the specific Env proteins expressed on the surface of the virion.

Infectious molecular clones

(IMCs). Full-length DNA clones of HIV-1 genomes capable of producing replication- competent viruses.

Cytotoxic T lymphocyte (CTL)-escape mutants

Viral variants that carry mutations in viral proteins that allow viruses to avoid detection by CTL. These mutations can reduce viral replicative fitness.

Interferon-α

(IFNα). A cytokine whose expression is rapidly upregulated after virus exposure to signal cells to create an antiviral environment. IFNα can reduce HIV-1 replication.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Joseph, S., Swanstrom, R., Kashuba, A. et al. Bottlenecks in HIV-1 transmission: insights from the study of founder viruses. Nat Rev Microbiol 13, 414–425 (2015). https://doi.org/10.1038/nrmicro3471

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro3471

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing